Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Oncotarget ; 12(8): 823-844, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33889304

ABSTRACT

Hypoxia stimulates neoangiogenesis, promoting tumor outgrowth, and triggers the epithelial-mesenchymal transition (EMT), which bestows cells with mesenchymal traits and multi-lineage differentiation potential. Here, we investigated whether EMT can confer endothelial attributes upon carcinoma cells, augmenting tumor growth and vascularization. Following orthotopic implantation of MCF-7 human epithelial breast cancer cells into mice, tumors of different sizes were immunostained for markers of hypoxia and EMT. Larger tumors were well-vascularized with CD31-positive cells of human origin. Hypoxic regions, demarcated by HIF-1α staining, exhibited focal areas of E-cadherin loss and elevated levels of vimentin and the EMT-mediator FOXC2. Implantation of MCF-7 cells, co-mixed with human mammary epithelial (HMLE) cells overexpressing the EMT-inducer Snail, markedly potentiated tumor growth and vascularization, compared with MCF-7 cells injected alone or co-mixed with HMLE-vector cells. Intra-tumoral vessels contained CD31-positive cells derived from either donor cell type. FOXC2 knockdown abrogated the potentiating effects of HMLE-Snail cells on MCF-7 tumor growth and vascularization, and compromised endothelial transdifferentiation of mesenchymal cells cultured in endothelial growth medium. Hence, cells that have undergone EMT can promote tumor growth and neovascularization either indirectly, by promoting endothelial transdifferentiation of carcinoma cells, or directly, by acquiring an endothelial phenotype, with FOXC2 playing key roles in these processes.

2.
J Clin Invest ; 130(6): 3315-3328, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32182215

ABSTRACT

The atypical cadherin FAT4 has established roles in the regulation of planar cell polarity and Hippo pathway signaling that are cell context dependent. The recent identification of FAT4 mutations in Hennekam syndrome, features of which include lymphedema, lymphangiectasia, and mental retardation, uncovered an important role for FAT4 in the lymphatic vasculature. Hennekam syndrome is also caused by mutations in collagen and calcium binding EGF domains 1 (CCBE1) and ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3), encoding a matrix protein and protease, respectively, that regulate activity of the key prolymphangiogenic VEGF-C/VEGFR3 signaling axis by facilitating the proteolytic cleavage and activation of VEGF-C. The fact that FAT4, CCBE1, and ADAMTS3 mutations underlie Hennekam syndrome suggested that all 3 genes might function in a common pathway. We identified FAT4 as a target gene of GATA-binding protein 2 (GATA2), a key transcriptional regulator of lymphatic vascular development and, in particular, lymphatic vessel valve development. Here, we demonstrate that FAT4 functions in a lymphatic endothelial cell-autonomous manner to control cell polarity in response to flow and is required for lymphatic vessel morphogenesis throughout development. Our data reveal a crucial role for FAT4 in lymphangiogenesis and shed light on the mechanistic basis by which FAT4 mutations underlie a human lymphedema syndrome.


Subject(s)
Cadherins/metabolism , Cell Polarity , Endothelial Cells/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Animals , Cadherins/genetics , Endothelial Cells/pathology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Humans , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/metabolism , Lymphedema/pathology , Mice , Mice, Transgenic , Syndrome
3.
Congenit Anom (Kyoto) ; 57(1): 24-31, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27783871

ABSTRACT

Foxc2, a member of the winged helix transcription factor family, is essential for eye, calvarial bone, cardiovascular and kidney development in mice. Nevertheless, how Foxc2-expressing cells and their descendent cells contribute to the development of these tissues and organs has not been elucidated. Here, we generated a Foxc2 knock-in (Foxc2CreERT2 ) mouse, in which administration of estrogen receptor antagonist tamoxifen induces nuclear translocation of Cre recombinase in Foxc2-expressing cells. By crossing with ROSA-LacZ reporter mice (Foxc2CreERT2 ; R26R), the fate of Foxc2 positive (Foxc2+ ) cells was analyzed through LacZ staining at various embryonic stages. We found Foxc2+ cell descendants in the supraoccipital and exoccipital bone in E18.5 embryos, when tamoxifen was administered at embryonic day (E) 8.5. Furthermore, Foxc2+ descendant cranial neural crest cells at E8-10 were restricted to the corneal mesenchyme, while Foxc2+ cell derived cardiac neural crest cells at E6-12 were found in the aorta, pulmonary trunk and valves, and endocardial cushions. Foxc2+ cell descendant contributions to the glomerular podocytes in the kidney were also observed following E6.5 tamoxifen treatment. Our results are consistent with previous reports of Foxc2 expression during early embryogenesis and the Foxc2CreERT2 mouse provides a tool to investigate spatiotemporal roles of Foxc2 and contributions of Foxc2+ expressing cells during mouse embryogenesis.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Organogenesis/genetics , Animals , Cell Lineage/genetics , Forkhead Transcription Factors/metabolism , Gene Order , Gene Targeting/methods , Genetic Loci , Genetic Vectors/genetics , Homologous Recombination , Immunohistochemistry , Kidney/embryology , Kidney/metabolism , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Transgenic
4.
Oncotarget ; 7(42): 68792-68802, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27634875

ABSTRACT

Osteosarcoma is a highly malignant tumor that contains a small subpopulation of tumor-propagating cells (also known as tumor-initiating cells) characterized by drug resistance and high metastatic potential. The molecular mechanism by which tumor-propagating cells promote tumor growth is poorly understood. Here, we report that the transcription factor forkhead box C2 (FOXC2) is frequently expressed in human osteosarcomas and is important in maintaining osteosarcoma cells in a stem-like state. In osteosarcoma cell lines, we show that anoikis conditions stimulate FOXC2 expression. Downregulation of FOXC2 decreases anchorage-independent growth and invasion in vitro and lung metastasis in vivo, while overexpression of FOXC2 increases tumor propagation in vivo. In osteosarcoma cell lines, we demonstrate that high levels of FOXC2 are associated with and required for the expression of osteosarcoma tumor-propagating cell markers. In FOXC2 knockdown cell lines, we show that CXCR4, a downstream target of FOXC2, can restore osteosarcoma cell invasiveness and metastasis to the lung.


Subject(s)
Bone Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Lung Neoplasms/secondary , Osteosarcoma/pathology , Receptors, CXCR4/metabolism , Animals , Anoikis , Cell Adhesion , Cell Differentiation , Cell Line, Tumor , Cell Movement , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Signal Transduction
5.
Data Brief ; 6: 514-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26904714

ABSTRACT

This data article shows the expression levels of specific podocyte injury markers and podocyte slit diaphragm protein nephrin in obese and lean Zucker rat glomeruli. It also contains information on the effect of the overexpression of transcription factor FOXC2 on the ratio of F- and G-actin and the expression level of ZO-1 in differentiated human podocytes. The article also shows data on the effect of treatments of differentiated podocytes with various factors associated with obesity and diabetes on the expression level of FOXC2. The detailed interpretation of these data and other aspects of podocyte injury mediated by upregulation of FOXC2 can be found in "Overexpression of transcription factor FOXC2 in cultured human podocytes upregulates injury markers and increases motility [1].

6.
Eur Heart J ; 37(18): 1469-75, 2016 May 07.
Article in English | MEDLINE | ID: mdl-26429810

ABSTRACT

AIM: Ventricular fibrillation (VF), the main cause of sudden cardiac death (SCD), occurs most frequently in the acute phase of myocardial infarction: a certain fraction of VF, however, develops in an apparently healthy heart, referred as idiopathic VF. The contribution of perturbation in the fast conduction system in the ventricle, the His-Purkinje system, for idiopathic VF has been implicated, but the underlying mechanism remains unknown. Irx3/IRX3 encodes a transcription factor specifically expressed in the His-Purkinje system in the heart. Genetic deletion of Irx3 provides a mouse model of ventricular fast conduction disturbance without anatomical or contraction abnormalities. The aim of this study was to examine the link between perturbed His-Purkinje system and idiopathic VF in Irx3-null mice, and to search for IRX3 genetic defects in idiopathic VF patients in human. METHODS AND RESULTS: Telemetry electrocardiogram recording showed that Irx3-deleted mice developed frequent ventricular tachyarrhythmias mostly at night. Ventricular tachyarrhythmias were enhanced by exercise and sympathetic nerve activation. In human, the sequence analysis of IRX3 exons in 130 probands of idiopathic VF without SCN5A mutations revealed two novel IRX3 mutations, 1262G>C (R421P) and 1453C>A (P485T). Ventricular fibrillation associated with physical activities in both probands with IRX3 mutations. In HL-1 cells and neonatal mouse ventricular myocytes, IRX3 transfection up-regulated SCN5A and connexin-40 mRNA, which was attenuated by IRX3 mutations. CONCLUSION: IRX3 genetic defects and resultant functional perturbation in the His-Purkinje system are novel genetic risk factors of idiopathic VF, and would improve risk stratification and preventive therapy for SCD in otherwise healthy hearts.


Subject(s)
Arrhythmias, Cardiac , Animals , Death, Sudden, Cardiac , Heart Conduction System , Homeodomain Proteins , Humans , Mice , Transcription Factors , Ventricular Fibrillation
7.
Exp Cell Res ; 340(1): 32-42, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26524507

ABSTRACT

Obesity and diabetes-related kidney diseases associate with renal failure and cardiovascular morbidity, and represent a major health issue worldwide. However, the molecular mechanisms leading to their development remain poorly understood. We observed increased expression of transcription factor FoxC2 in the podocytes of obese Zucker rats that are insulin resistant and albuminuric. We also found that depletion of adiponectin, an adipocyte-derived hormone whose secretion is decreased in obesity, upregulated FOXC2 in differentiated human podocytes in vitro. Overexpression of FOXC2 in cultured human podocytes led to increased nuclear expression of FOXC2 associated with a change of cellular morphology. This was accompanied by upregulation of vimentin, a key mesenchymal marker, and active beta-catenin, associated with podocyte injury. We also observed re-organization of the actin cytoskeleton, disrupted localization of the tight junction protein ZO-1, and increased motility of podocytes overexpressing FOXC2. These data indicate that the expression of FOXC2 in podocytes needs to be tightly regulated, and that its overexpression induces a chain of cellular events leading to podocyte dysfunction. These changes may lead to podocyte detachment and depletion ultimately contributing to albuminuria. We also suggest a novel molecular mechanism linking obesity-induced decrease in adiponectin to podocyte dysfunction via upregulation of FOXC2.


Subject(s)
Cell Movement , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Podocytes/metabolism , Podocytes/pathology , Up-Regulation , Animals , Biomarkers/metabolism , Cells, Cultured , Forkhead Transcription Factors/biosynthesis , Humans , Rats , Rats, Zucker , Wound Healing
8.
Dev Biol ; 409(1): 218-233, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26542011

ABSTRACT

Lymph is returned to the blood circulation exclusively via four lymphovenous valves (LVVs). Despite their vital importance, the architecture and development of LVVs is poorly understood. We analyzed the formation of LVVs at the molecular and ultrastructural levels during mouse embryogenesis and identified three critical steps. First, LVV-forming endothelial cells (LVV-ECs) differentiate from PROX1(+) progenitors and delaminate from the luminal side of the veins. Second, LVV-ECs aggregate, align perpendicular to the direction of lymph flow and establish lympho-venous connections. Finally, LVVs mature with the recruitment of mural cells. LVV morphogenesis is disrupted in four different mouse models of primary lymphedema and the severity of LVV defects correlate with that of lymphedema. In summary, we have provided the first and the most comprehensive analysis of LVV development. Furthermore, our work suggests that aberrant LVVs contribute to lymphedema.


Subject(s)
Lymphatic Vessels/embryology , Lymphedema/embryology , Lymphedema/pathology , Venous Valves/embryology , Animals , Animals, Newborn , Cell Differentiation , Disease Models, Animal , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Lymphatic Vessels/ultrastructure , Mice, Inbred C57BL , Morphogenesis , Penetrance , Phenotype , Venous Valves/ultrastructure
9.
J Clin Invest ; 125(10): 3861-77, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26389677

ABSTRACT

Biomechanical forces, such as fluid shear stress, govern multiple aspects of endothelial cell biology. In blood vessels, disturbed flow is associated with vascular diseases, such as atherosclerosis, and promotes endothelial cell proliferation and apoptosis. Here, we identified an important role for disturbed flow in lymphatic vessels, in which it cooperates with the transcription factor FOXC2 to ensure lifelong stability of the lymphatic vasculature. In cultured lymphatic endothelial cells, FOXC2 inactivation conferred abnormal shear stress sensing, promoting junction disassembly and entry into the cell cycle. Loss of FOXC2-dependent quiescence was mediated by the Hippo pathway transcriptional coactivator TAZ and, ultimately, led to cell death. In murine models, inducible deletion of Foxc2 within the lymphatic vasculature led to cell-cell junction defects, regression of valves, and focal vascular lumen collapse, which triggered generalized lymphatic vascular dysfunction and lethality. Together, our work describes a fundamental mechanism by which FOXC2 and oscillatory shear stress maintain lymphatic endothelial cell quiescence through intercellular junction and cytoskeleton stabilization and provides an essential link between biomechanical forces and endothelial cell identity that is necessary for postnatal vessel homeostasis. As FOXC2 is mutated in lymphedema-distichiasis syndrome, our data also underscore the role of impaired mechanotransduction in the pathology of this hereditary human disease.


Subject(s)
Endothelial Cells/cytology , Forkhead Transcription Factors/physiology , Lymphatic System/growth & development , Lymphatic Vessels/cytology , Rheology , Acyltransferases , Adaptor Proteins, Signal Transducing/physiology , Animals , Apoptosis , Cell Cycle , Cell Division , Cells, Cultured , Cytoskeleton/ultrastructure , Endothelial Cells/pathology , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/deficiency , Humans , Intercellular Junctions/ultrastructure , Lymphatic Vessels/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/physiology , RNA Interference , RNA, Small Interfering/pharmacology , Stress Fibers/ultrastructure , Stress, Mechanical , Transcription Factors/physiology , Transcription, Genetic , Transfection , YAP-Signaling Proteins
10.
Biomed Res ; 36(4): 235-45, 2015.
Article in English | MEDLINE | ID: mdl-26299482

ABSTRACT

The forkhead box C2 (Foxc2) protein is a member of the forkhead/winged helix transcription factor family and plays an essential role in cardiovascular development. Previous studies showed that Foxc2 null mouse embryos die during midgestation or just after birth with severe cardiovascular defects, including interruption, coarctation of the aortic arch and ventricular septal defects. These are also seen in human congenital heart disease. However, the tissue specific role of Foxc2 in aortic arch remodelling is not yet fully understood. Here we show that Foxc2 is expressed in a restricted pattern in several cell populations, including the mesenchyme and endothelium of pharyngeal arch arteries, which are important for cardiovascular development. In this study, we use a conditional knockout approach to examine the tissue specific role of Foxc2 in aortic arch remodelling. We demonstrate that mouse embryos lacking Foxc2 in Nkx2.5-expressing mesenchyme and endothelium of pharyngeal arch arteries display aortic arch interruption type B and ventricular septal defects. In contrast, conditional deletion of Foxc2 in Tie2-expressing endothelial cells does not result in aortic arch or ventricular septal defects, but does result in embryonic lethality due to peripheral oedema. Our data therefore provide for a detailed understanding of the role of mesenchymal Foxc2 in aortic arch remodelling and in the development of ventricular septum.


Subject(s)
Aorta, Thoracic/embryology , Aorta, Thoracic/metabolism , Branchial Region/metabolism , Forkhead Transcription Factors/genetics , Mesoderm/metabolism , Ventricular Septum/metabolism , Alleles , Animals , Branchial Region/embryology , Cardiovascular Abnormalities/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Gene Targeting , Genotype , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesoderm/embryology , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Ventricular Septum/embryology
11.
J Clin Invest ; 125(8): 2979-94, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26214525

ABSTRACT

Heterozygous germline mutations in the zinc finger transcription factor GATA2 have recently been shown to underlie a range of clinical phenotypes, including Emberger syndrome, a disorder characterized by lymphedema and predisposition to myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). Despite well-defined roles in hematopoiesis, the functions of GATA2 in the lymphatic vasculature and the mechanisms by which GATA2 mutations result in lymphedema have not been characterized. Here, we have provided a molecular explanation for lymphedema predisposition in a subset of patients with germline GATA2 mutations. Specifically, we demonstrated that Emberger-associated GATA2 missense mutations result in complete loss of GATA2 function, with respect to the capacity to regulate the transcription of genes that are important for lymphatic vessel valve development. We identified a putative enhancer element upstream of the key lymphatic transcriptional regulator PROX1 that is bound by GATA2, and the transcription factors FOXC2 and NFATC1. Emberger GATA2 missense mutants had a profoundly reduced capacity to bind this element. Conditional Gata2 deletion in mice revealed that GATA2 is required for both development and maintenance of lymphovenous and lymphatic vessel valves. Together, our data unveil essential roles for GATA2 in the lymphatic vasculature and explain why a select catalogue of human GATA2 mutations results in lymphedema.


Subject(s)
GATA2 Transcription Factor/metabolism , Lymphatic Vessels/embryology , Lymphedema/embryology , Mutation , Animals , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/genetics , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/pathology , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Nat Commun ; 6: 7274, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26027726

ABSTRACT

The lymphatic system maintains tissue fluid balance, and dysfunction of lymphatic vessels and valves causes human lymphedema syndromes. Yet, our knowledge of the molecular mechanisms underlying lymphatic vessel development is still limited. Here, we show that cyclin-dependent kinase 5 (Cdk5) is an essential regulator of lymphatic vessel development. Endothelial-specific Cdk5 knockdown causes congenital lymphatic dysfunction and lymphedema due to defective lymphatic vessel patterning and valve formation. We identify the transcription factor Foxc2 as a key substrate of Cdk5 in the lymphatic vasculature, mechanistically linking Cdk5 to lymphatic development and valve morphogenesis. Collectively, our findings show that Cdk5-Foxc2 interaction represents a critical regulator of lymphatic vessel development and the transcriptional network underlying lymphatic vascular remodeling.


Subject(s)
Cyclin-Dependent Kinase 5/genetics , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Animals , Cyclin-Dependent Kinase 5/metabolism , Endothelial Cells/cytology , Forkhead Transcription Factors/genetics , Human Umbilical Vein Endothelial Cells , Humans , Lymphatic Vessels/pathology , Mice , Mice, Knockout , Phosphorylation , Real-Time Polymerase Chain Reaction , Stress, Mechanical
13.
Biomed Res ; 36(3): 159-67, 2015.
Article in English | MEDLINE | ID: mdl-26106045

ABSTRACT

Patients chronically infected with hepatitis C virus (HCV) are at risk of developing end-stage liver disease and hepatocellular carcinoma. Development of drugs to inhibit hepatocyte damage and a vaccine against HCV is hampered by the lack of a small animal model. We generated mice in which the viral genome RNA was always present in the hepatocytes using a special transgene. Here we show that the HCV genome RNA transcribed by Pol I polymerase can replicate and produce infectious viruses in mice. We obtained a transgenic mouse with 200 copies per haploid which we named the A line mouse. It produced ~ 3 × 10(6) HCV RNA copies/mL serum, which is at the comparable level as patients with chronic HCV infection. This mouse was immunotolerant to HCV and showed hepatic steatosis without any necroinflammation at the age of 6 months or hepatocellular carcinoma at the age of 15 months. Thus, the A line mouse can be used as an animal model for chronic HCV infection. This will enable better study of the abnormalities in metabolism and signal transduction in infected hepatocytes, and development of drugs that cure abnormalities.


Subject(s)
Fatty Liver/etiology , Genome, Viral , Hepacivirus/physiology , Hepatitis C/complications , Hepatitis C/virology , RNA, Viral , Virus Replication , Animals , Disease Models, Animal , Fatty Liver/pathology , Humans , Mice , Mice, Transgenic , RNA Polymerase I/metabolism , Time Factors , Transcription, Genetic , Viral Load
14.
Biol Pharm Bull ; 38(6): 935-40, 2015.
Article in English | MEDLINE | ID: mdl-25810454

ABSTRACT

The ubiquitin-proteasome pathway plays an important role in regulating apoptosis and the cell cycle. Recently, proteasome inhibitors have been shown to have antitumor effects and have been used in anticancer therapy for several cancers such as multiple myeloma. Although some flavones, such as apigenin, chrysin and luteolin, have a specific role in the inhibition of proteasome activity and induced apoptosis in some reports, these findings did not address all flavone types. To further investigate the proteasome-inhibitory mechanism of flavonoids, we examined the inhibitory activity of 5,6,7-trihydroxyflavone, baicalein and 5,6,7,4'-tetrahydroxyflavone, scutellarein on extracted proteasomes from mice and cancer cells. Unlike the other flavones, baicalein and scutellarein did not inhibit proteasome activity or accumulate levels of ubiquitinated proteins. These results indicate that flavones with hydroxy groups at positions 5, 6 and 7 of the A-ring lack the anti-proteasome function.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents/pharmacology , Flavones/pharmacology , Neoplasms/metabolism , Plant Extracts/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Apigenin/chemistry , Apigenin/pharmacology , Apoptosis , Flavanones/chemistry , Flavanones/pharmacology , Flavones/chemistry , HCT116 Cells , Humans , Jurkat Cells , Molecular Structure , Phytotherapy , Plant Extracts/chemistry , Proteasome Inhibitors/chemistry , Rabbits , Structure-Activity Relationship
15.
Dev Biol ; 386(1): 25-33, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24361262

ABSTRACT

During embryogenesis, lymphatic endothelial progenitor cells first arise from a subset of blood vascular endothelial cells in the dorsolateral aspects of the cardinal veins. The molecular cues responsible for defining the regionalisation of such a discrete pool of progenitors remain uncharacterised. Here we identify a novel function for CYP26B1, an enzyme known to play a role in tissue morphogenesis by fine-tuning retinoic acid (RA) concentration, in regulating lymphangiogenesis. Cyp26b1-null mice, in which RA levels are elevated, exhibited an increased number of lymphatic endothelial progenitor cells in the cardinal veins, together with hyperplastic, blood filled lymph sacs and hyperplastic dermal lymphatic vessels. Conversely, mice over-expressing Cyp26b1 had hypoplastic lymph sacs and lymphatic vessels. Our data suggest that RA clearance by CYP26B1 in the vicinity of lymphatic endothelial progenitor cells is important for determining the position and size of the progenitor pool specified. Our studies identify a genetic pathway that underpins the architecture of the developing lymphatics and define CYP26B1 as a novel modulator of lymphatic vascular patterning.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Developmental , Lymphangiogenesis , Lymphatic System/embryology , Lymphatic Vessels/metabolism , Retinoids/metabolism , Animals , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Endothelial Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Phenotype , Retinoic Acid 4-Hydroxylase , Signal Transduction , Transgenes , Tretinoin/metabolism
16.
Mol Cell Biol ; 33(19): 3749-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878394

ABSTRACT

One of the key mechanisms linking cell signaling and control of gene expression is reversible phosphorylation of transcription factors. FOXC2 is a forkhead transcription factor that is mutated in the human vascular disease lymphedema-distichiasis and plays an essential role in lymphatic vascular development. However, the mechanisms regulating FOXC2 transcriptional activity are not well understood. We report here that FOXC2 is phosphorylated on eight evolutionarily conserved proline-directed serine/threonine residues. Loss of phosphorylation at these sites triggers substantial changes in the FOXC2 transcriptional program. Through genome-wide location analysis in lymphatic endothelial cells, we demonstrate that the changes are due to selective inhibition of FOXC2 recruitment to chromatin. The extent of the inhibition varied between individual binding sites, suggesting a novel rheostat-like mechanism by which expression of specific genes can be differentially regulated by FOXC2 phosphorylation. Furthermore, unlike the wild-type protein, the phosphorylation-deficient mutant of FOXC2 failed to induce vascular remodeling in vivo. Collectively, our results point to the pivotal role of phosphorylation in the regulation of FOXC2-mediated transcription in lymphatic endothelial cells and underscore the importance of FOXC2 phosphorylation in vascular development.


Subject(s)
Endothelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Transcription, Genetic/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Cells, Cultured , Chlorocebus aethiops , Forkhead Transcription Factors/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Immunoblotting , Mice , Mice, Transgenic , Microscopy, Confocal , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Phosphorylation , Proline/genetics , Proline/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism
17.
Cancer Res ; 73(6): 1981-92, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378344

ABSTRACT

Resistance to chemotherapy and metastases are the major causes of breast cancer-related mortality. Moreover, cancer stem cells (CSC) play critical roles in cancer progression and treatment resistance. Previously, it was found that CSC-like cells can be generated by aberrant activation of epithelial-mesenchymal transition (EMT), thereby making anti-EMT strategies a novel therapeutic option for treatment of aggressive breast cancers. Here, we report that the transcription factor FOXC2 induced in response to multiple EMT signaling pathways as well as elevated in stem cell-enriched factions is a critical determinant of mesenchymal and stem cell properties, in cells induced to undergo EMT- and CSC-enriched breast cancer cell lines. More specifically, attenuation of FOXC2 expression using lentiviral short hairpin RNA led to inhibition of the mesenchymal phenotype and associated invasive and stem cell properties, which included reduced mammosphere-forming ability and tumor initiation. Whereas, overexpression of FOXC2 was sufficient to induce CSC properties and spontaneous metastasis in transformed human mammary epithelial cells. Furthermore, a FOXC2-induced gene expression signature was enriched in the claudin-low/basal B breast tumor subtype that contains EMT and CSC features. Having identified PDGFR-ß to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Thus, FOXC2 or its associated gene expression program may provide an effective target for anti-EMT-based therapies for the treatment of claudin-low/basal B breast tumors or other EMT-/CSC-enriched tumors.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Transformed , Female , Forkhead Transcription Factors/genetics , Gene Expression , Humans , Mice , Mice, Inbred NOD , Mice, SCID
18.
Transgenic Res ; 22(3): 659-66, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23117587

ABSTRACT

Thymus development is a complicated process that includes highly dynamic morphological changes and reciprocal tissue interactions between endoderm-derived epithelial cells of the anterior foregut and neural crest-derived mesenchymal cells. We generated and characterized a Tbx1-AmCyan1 reporter transgenic mouse to visualize thymus precursor cells during early embryonic development. In transgenic embryos, AmCyan1 fluorescence was specifically detected in the endoderm of the developing 3rd and 4th pharyngeal pouches and later in thymus epithelium until E14.5. Cells expressing AmCyan1 that were isolated based on AmCyan1 fluorescence expressed endodermal, thymic, and parathyroid markers, but they did not express neural crest or endothelial markers; these findings indicated that this transgenic mouse strain could be used to collect thymic or parathyroid precursor cells or both. We also showed that in nude mice, which exhibit defects in thymus development, the thymus precursors were clearly labeled with AmCyan1. In summary, these AmCyan1-fluorescent transgenic mice are useful for investigating early thymus development.


Subject(s)
Mice, Transgenic , Thymus Gland/cytology , Thymus Gland/embryology , Animals , Biomarkers/metabolism , Endoderm/cytology , Endoderm/embryology , Female , Genes, Reporter , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Neural Crest/metabolism , Rabbits , Rats , T-Box Domain Proteins/genetics
19.
Phytother Res ; 27(9): 1362-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23147714

ABSTRACT

Baicalin and scutellarin are the major active principal flavonoids extracted from the Chinese herbal medicines Scutellaria baicalensis and Erigeron breviscapus (Vant.) Hand-Mazz. It has recently been reported that baicalin and scutellarin have antitumor activity. However, the mechanisms of action are unknown. We previously reported that some flavonoids have a specific role in the inhibition of the activity of proteasome subunits and induced apoptosis in tumor cells. To further investigate these pharmacological effects, we examined the inhibitory activity of baicalin and scutellarin on the extracted proteasomes from mice and cancer cells. Using fluorogenic substrates for proteasome catalytic subunits, we found that baicalin and scutellarin specifically inhibited chymotrypsin-like activity but did not inhibit trypsin-like and peptidyl-glutamyl peptide hydrolyzing activities. These data suggested that baicalin and scutellarin specifically inhibit chymotrypsin-like catalytic activity in the proteasome.


Subject(s)
Apigenin/pharmacology , Chymotrypsin/antagonists & inhibitors , Flavonoids/pharmacology , Glucuronates/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival , Erigeron/chemistry , Female , Humans , Liver/enzymology , Mice , Mice, Inbred ICR , Proteasome Endopeptidase Complex/drug effects , Scutellaria baicalensis/chemistry
20.
Dev Dyn ; 241(11): 1744-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22972661

ABSTRACT

BACKGROUND: In previous studies, we investigated the effects of excess retinoic acid (RA) during palatogenesis by RA administration to pregnant mice. In the present study, we deleted Cyp26b1, one of the RA-degrading enzymes, to further study the effects of excess RA in the normal developing palate and to understand how endogenous levels of RA are regulated. RESULTS: Excess RA, due to the absence of Cyp26b1, targets cells in the bend region of the palatal shelves and inhibits their horizontal elevation, leading to cleft palate. An organ culture of Cyp26b1-/- palatal shelves after tongue removal did not rescue the impaired elevation of the palatal shelves. The expression of Fgf10, Bmp2, and Tbx1, important molecules in palatal development, was down-regulated. Cell proliferation was decreased in the bend region of palatal shelves. Tongue muscles were hypoplastic and/or missing in Cyp26b1-/- mice. CONCLUSIONS: We demonstrated that CYP26B1 is essential during palatogenesis. Excess RA due to the lack of Cyp26b1 suppresses the expression of key regulators of palate development in the bend region, resulting in a failure in the horizontal elevation of the palatal shelves. The regulation of RA signaling through CYP26B1 is also necessary for the development of tongue musculature and for tongue depression.


Subject(s)
Apoptosis/physiology , Cytochrome P-450 Enzyme System/metabolism , Palate/embryology , Palate/metabolism , Tretinoin/metabolism , Animals , Apoptosis/genetics , Cell Proliferation , Cytochrome P-450 Enzyme System/genetics , Female , Mice , Mice, Knockout , Organ Culture Techniques , Pregnancy , Real-Time Polymerase Chain Reaction , Retinoic Acid 4-Hydroxylase
SELECTION OF CITATIONS
SEARCH DETAIL
...